SUPPLEMENTAL METHODS, FIGURES and TABLES Cohesin Mutations Alter DNA Damage Repair and Chromatin Structure and Create Therapeuti

Total Page:16

File Type:pdf, Size:1020Kb

SUPPLEMENTAL METHODS, FIGURES and TABLES Cohesin Mutations Alter DNA Damage Repair and Chromatin Structure and Create Therapeuti SUPPLEMENTAL METHODS, FIGURES and TABLES Cohesin mutations alter DNA damage repair and chromatin structure and create therapeutic vulnerabilities in MDS/AML Zuzana Tothova1,2*, Anne-Laure Valton3, Rebecca A. Gorelov2, Mounica Vallurupalli1,2, John M. Krill-Burger2, Amie Holmes4, Catherine C. LanDers2, J. Erika HayDu2, EDyta Malolepsza2, Christina Hartigan2, Melanie Donahue2, Katerina D. Popova2, Sebastian Koochaki1, Sergey V. Venev3, Jeanne Rivera5, EDwin Chen5, Kasper Lage2, Monica Schenone2, Alan D. D'AnDrea4, Steven A. Carr2, Elizabeth A. Morgan6, Job Dekker3,7, Benjamin L. Ebert1,2,7* 1 SUPPLEMENTAL METHODS Generation of cohesin-mutant single cell clones Both U937 anD K562 parental cell lines were confirmeD to be wilD type for all cohesin complex components and regulators based on RNA-Seq, anD exome or genome sequencing Data generated by the Cancer Cell Line Encyclopedia (CCLE) prior to development of any cohesin- mutant moDels(1). Of note, since STAG2 is an X-linkeD gene which unDergoes normal X- inactivation in females, patients with preDicted loss-of-function (LOF) STAG2 mutations are predicted to lack normal STAG2 expression. Therefore, in both cases of U937 cells and K562 cells, which both carry two copies of the X chromosome, we screened for presence of homozygous STAG2 LOF mutations. U937- and K562-Cas9 expressing cells were first generateD by lentiviral transduction of parental U937 and K562 cells with a lentiviral vector expressing the Cas9 nuclease unDer blasticiDin selection (pLX-311Cas9; Addgene #96924). Each Cas9- expressing cell line was subjected to a Cas9 activity assay using a separate lentiviral transduction with a GFP reporter construct (pXPR-011; Addgene #59702) and only cell lines with >80% Cas9 activity as determined by flow cytometry were used for subsequent generation of single cell clones. To generate the single cell clones, we cloned sgRNAs targeting STAG2, SMC3, RAD21 or non-targeting sgRNAs into a minimal backbone plasmiD (ADDgene #41824) and transfected into U937-Cas9 or K562-Cas9 cells using nucleofection (Lonza, Nucleofector II). We single cell sorted GFP+ cells into 96 well plates 36-48 hours after transfection, and grew up single cell clones, which we confirmed by DNA sequencing of the targeted locus, as well as Western blotting. Select cohesin-mutant clones were transDuceD with GFP or mCherry anD luciferase containing lentiviral plasmids (lenti-GFP-Luc2, or lenti-mCherry-Luc2) and sorted for GFP or mCherry positivity for use in mouse transplant experiments and competition assays. Choice of sgRNAs was Driven by observation of presumeD pathogenic STAG2 mutations in patients baseD on RapiD Heme Panel sequencing at the Dana-Farber Cancer Institute (DFCI cBioPortal) (2). Western Blotting and Immunoprecipitation (IP) Cells were washeD in colD 1X PBS (Corning CellGro) anD lyseD for 15-30min on ice using NP-40 lysis buffer (150mM NaCl, 50mM Tris pH 7.5 (Thermo Fisher Scientific 15567027), 1% NP40 (Roche 11332473001), 5% glycerol (Sigma-AlDrich G5516), anD 100X Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific 78447) in water). Lysates were quantified using Pierce BCA Protein Assay Kit (Thermo Fisher Scientific 23225) and reduced with 6X Laemmli SDS-Sample Buffer (Boston BioProDucts BP-111R) and 10X NuPAGE Sample ReDucing Agent (Thermo Fisher Scientific NP0009) for 5min at 95°C. For SMC1A IPs, protein lysates were incubateD with SMC1A antiboDy overnight rotating at 4°C. 50µL of Protein G or A agarose beads in a 50% slurry (EZview Red Protein G Affinity Gel, Sigma- AlDrich, E3403 anD EZview ReD Protein A Affinity Gel, Sigma-AlDrich, P6486) were washeD three times with NP-40 lysis buffer and incubated with the antibody and lysate mixture. The mixture was rotateD for 2hrs at 4°C, anD the beaDs were then washed once with NP-40 lysis buffer and twice with wash buffer (150mM NaCl, 50mM Tris pH 7.5 (Thermo Fisher Scientific 15567027), 1% glycerol (Sigma-AlDrich G5516), 100X Halt Protease anD Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific 78447) in water). Samples were reDuceD as stateD above. For Western blotting, samples were run on NuPAGE 4-12% Bis-Tris Protein Gels (Invitrogen NP0321 anD NP0316) in NuPAGE MOPS SDS Running Buffer 20X (Life Technologies NP0001) or NuPAGE MES SDS Running Buffer 20X (Life Technologies NP0002) and transferred onto Nitrocellulose Pre-Cut Blotting Membranes (0.45µm, Invitrogen LC2001) using 10X Tris/Glycine 2 Buffer (Bio-RaD 1610734) anD 20% Methanol (VWR TXBJ349664LBRI) in water. Gels were transferreD at 90V for 2-2.5hrs or 40V overnight at 4°C. Membranes were blocked with 5% non- fat Dry milk (Bio-RaD 1706404XTU) in Tris BuffereD Saline with Tween-20 (Cell Signaling Technology, 9997) for 30-60min at room temperature. Incubation with primary antibody was performed overnight at 4°C or for 2-4hrs at room temperature in 5% milk in TBST. Primary antibodies used for characterization of control and STAG2 knockout clones are as follows: Phospho-Histone H2A.X, STAG2, anti-SA1, anD anti-SMC1. Membranes were washeD 3 times for 5min each in TBST and incubated in Anti-rabbit IgG, Anti-mouse IgG, or Rabbit anti-Goat IgG secondary as needed in 5% milk in TBST for 1hr at room temperature. Membranes were washed an additional 3 times for 5min each in TBST and visualized after a 5min incubation with SuperSignal West Dura ExtenDeD Duration Substrate (Thermo Scientific, 34075), SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific, 34078), or Amersham ECL Prime Western Blotting Detection Reagent (GE Healthcare, RPN2232). AdDitional blots were imageD using the LI-COR platform (LI-COR Biosciences) with IRDye 800CW anD IRDye680RD secondary antibodies (mouse, IRDye 680LT Donkey-anti-mouse IgG(925-68022) used at 1:15,000; rabbit, IRDye 800CW Goat anti-rabbit IgG (926-32211) used at 1:10,000) and imaging was performeD on an ODyssey CLx InfrareD Imaging System. To assess levels of DNA Damage, cells were treateD with 100nM Talazoparib (Selleck S7048) and incubated at 37°C for 24hrs before harvesting protein lysate. Cells were also irraDiateD using an X-ray irraDiator at 100raDs or 10Gy, anD protein lysate was harvesteD after 1hr at 37°C. Samples were run on NuPAGE 3-8% Tris-Acetate Protein Gels (Thermo Fisher Scientific, EA0375) anD transferreD as above at 90V for 2hrs at 4°C. Membranes were blockeD for 30min at room temperature anD incubateD with the primary antiboDy overnight at 4°C. Primary antiboDies used are as follows: ATR, Chk1, Phospho-Chk2, anD Vinculin in 5% milk in TBST; Anti-Chk2 in 1% milk in TBST; Phospho-ATR, ATM , Phospho-ATM, anD Phospho-Chk1 in 5% BSA in TBST. Membranes were washeD 10 times for 10min each anD incubateD with the seconDary antiboDy as stated above for 1hr at room temperature. After incubation, membranes were washed an additional 10 times for 10min each and visualized as above. The following antiboDies were useD in the stuDy: Anti-ATR (Abcam, Cat#ab54793) Anti-SA1 (Abcam, Cat#ab4455) Anti-SMC1 (Abcam, Cat#ab140493) STAG2 (Cell Signaling Technology, Cat#4239) β-Actin, 8H10D10 (Cell Signaling Technology, Cat#3700) Phospho-Histone H2A.X, Ser139, 20E3 (Cell Signaling Technology, Cat#9718) Chk1, 2G1D5 (Cell Signaling Technology, Cat#2360) Phospho-Chk2, Th468, C13C1 (Cell Signaling Technology, Cat#2197) Phospho-ATR, Ser428, D6H9 (Cell Signaling Technology, Cat#2853) ATM, 11G12 (Cell Signaling Technology, Cat#92356) Phospho-ATM, Ser1981, D6H9 (Cell Signaling Technology, Cat#5883) Phospho-Chk1, ser345, 133D3 (Cell Signaling Technology, Cat#2348) Vinculin, E1E9V XP (Cell Signaling Technology, Cat#13901) Anti-rabbit IgG, HRP-linkeD (Cell Signaling Technology, Cat#7074) Anti-mouse IgG, HRP-linkeD (Cell Signaling Technology, Cat#7076) Anti-Chk2, clone 7 (EMD Millipore, Cat#05-649) Rabbit anti-Goat IgG, HRP conjugate (EMD Millipore, Cat#AP106P) 3 Genome-wide CRISPR screening 5 STAG2-mutant anD 6 STAG2-wilD type U937 cells expressing Cas9 anD confirmeD to have >80% Cas9 activity by the GFP reporter construct (pXPR-011; Addgene #59702) were infecteD with the genome-wiDe human Avana LentiGuiDe-Puro CRISPR library (BroaD Genetic Perturbations Platform), which contains ~75,000 sgRNAs targeting ~19,000 genes anD 1000 controls, in two separate experiments, as previously described (3). Briefly, cells were selecteD in blasticidin for 14 Days to maximize Cas9 expression. BlasticiDin was washeD out of the meDia two days prior to transduction. Puromycin selection of transduced cells was initiated on Day 2 after transDuction anD carrieD out for a total of 5 Days. BlasticiDin was restarteD on Day 8 after transDuction anD incluDeD in the meDia through the enD of the experiment. Cell pellets were collected on Day 21, genomic DNA was purified and the sgRNA sequences amplified with sufficient gDNA to maintain representation, and quantified using next generation sequencing. Transplantation assays and mouse analysis. 8-10 week old male and female NSGS mice (NOD-SCID; IL2Rγ null; Tg(IL3, CSF2, KITL)) obtained from The Jackson laboratory, strain 013062) were sublethally irradiated using Gamma- cell irradiator (Best Theratronics) at a dose of 250 Rads, and retro-orbitally injected with 200,000- 1 million cells/mouse. 4-10 recipients were injected per arm. Animals were monitored daily for presence of disease, and were sacrificed at designated times after transplantation or when moribunD. Peripheral blooD was collecteD from the retro-orbital cavity using a heparinized glass capillary and automated total and differential blood cell counts were determined using ADVIA Hematology system (Bayer). Collected blood was also used to prepare blood smears, which were stained with May Gruenwald and Modified Giemsa (Sigma Aldrich). Following sacrifice, mice were examined for presence of tumors, enlarged lymph nodes or other abnormalities, and organs were collecteD for further cell anD histopathologic analysis. Single cell suspensions were maDe from bone marrow, and spleen, washed, red blood cells were lysed, and samples were frozen in 10% dimethylsulfoxide/ 90% FCS until analysis.
Recommended publications
  • Mutational Inactivation of STAG2 Causes Aneuploidy in Human Cancer
    REPORTS mean difference for all rubric score elements was ing becomes a more commonly supported facet 18. C. L. Townsend, E. Heit, Mem. Cognit. 39, 204 (2011). rejected. Univariate statistical tests of the observed of STEM graduate education then students’ in- 19. D. F. Feldon, M. Maher, B. Timmerman, Science 329, 282 (2010). mean differences between the teaching-and- structional training and experiences would alle- 20. B. Timmerman et al., Assess. Eval. High. Educ. 36,509 research and research-only conditions indicated viate persistent concerns that current programs (2011). significant results for the rubric score elements underprepare future STEM faculty to perform 21. No outcome differences were detected as a function of “testability of hypotheses” [mean difference = their teaching responsibilities (28, 29). the type of teaching experience (TA or GK-12) within the P sample population participating in both research and 0.272, = 0.006; CI = (.106, 0.526)] with the null teaching. hypothesis rejected in 99.3% of generated data References and Notes 22. Materials and methods are available as supporting samples (Fig. 1) and “research/experimental de- 1. W. A. Anderson et al., Science 331, 152 (2011). material on Science Online. ” P 2. J. A. Bianchini, D. J. Whitney, T. D. Breton, B. A. Hilton-Brown, 23. R. L. Johnson, J. Penny, B. Gordon, Appl. Meas. Educ. 13, sign [mean difference = 0.317, = 0.002; CI = Sci. Educ. 86, 42 (2001). (.106, 0.522)] with the null hypothesis rejected in 121 (2000). 3. C. E. Brawner, R. M. Felder, R. Allen, R. Brent, 24. R. J. A. Little, J.
    [Show full text]
  • The Mutational Landscape of Myeloid Leukaemia in Down Syndrome
    cancers Review The Mutational Landscape of Myeloid Leukaemia in Down Syndrome Carini Picardi Morais de Castro 1, Maria Cadefau 1,2 and Sergi Cuartero 1,2,* 1 Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; [email protected] (C.P.M.d.C); [email protected] (M.C.) 2 Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain * Correspondence: [email protected] Simple Summary: Leukaemia occurs when specific mutations promote aberrant transcriptional and proliferation programs, which drive uncontrolled cell division and inhibit the cell’s capacity to differentiate. In this review, we summarize the most frequent genetic lesions found in myeloid leukaemia of Down syndrome, a rare paediatric leukaemia specific to individuals with trisomy 21. The evolution of this disease follows a well-defined sequence of events and represents a unique model to understand how the ordered acquisition of mutations drives malignancy. Abstract: Children with Down syndrome (DS) are particularly prone to haematopoietic disorders. Paediatric myeloid malignancies in DS occur at an unusually high frequency and generally follow a well-defined stepwise clinical evolution. First, the acquisition of mutations in the GATA1 transcription factor gives rise to a transient myeloproliferative disorder (TMD) in DS newborns. While this condition spontaneously resolves in most cases, some clones can acquire additional mutations, which trigger myeloid leukaemia of Down syndrome (ML-DS). These secondary mutations are predominantly found in chromatin and epigenetic regulators—such as cohesin, CTCF or EZH2—and Citation: de Castro, C.P.M.; Cadefau, in signalling mediators of the JAK/STAT and RAS pathways.
    [Show full text]
  • Redundant and Specific Roles of Cohesin STAG Subunits in Chromatin Looping and Transcriptional Control
    Downloaded from genome.cshlp.org on October 10, 2021 - Published by Cold Spring Harbor Laboratory Press Research Redundant and specific roles of cohesin STAG subunits in chromatin looping and transcriptional control Valentina Casa,1,6 Macarena Moronta Gines,1,6 Eduardo Gade Gusmao,2,3,6 Johan A. Slotman,4 Anne Zirkel,2 Natasa Josipovic,2,3 Edwin Oole,5 Wilfred F.J. van IJcken,1,5 Adriaan B. Houtsmuller,4 Argyris Papantonis,2,3 and Kerstin S. Wendt1 1Department of Cell Biology, Erasmus MC, 3015 GD Rotterdam, The Netherlands; 2Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany; 3Institute of Pathology, University Medical Center, Georg-August University of Göttingen, 37075 Göttingen, Germany; 4Optical Imaging Centre, Erasmus MC, 3015 GD Rotterdam, The Netherlands; 5Center for Biomics, Erasmus MC, 3015 GD Rotterdam, The Netherlands Cohesin is a ring-shaped multiprotein complex that is crucial for 3D genome organization and transcriptional regulation during differentiation and development. It also confers sister chromatid cohesion and facilitates DNA damage repair. Besides its core subunits SMC3, SMC1A, and RAD21, cohesin in somatic cells contains one of two orthologous STAG sub- units, STAG1 or STAG2. How these variable subunits affect the function of the cohesin complex is still unclear. STAG1- and STAG2-cohesin were initially proposed to organize cohesion at telomeres and centromeres, respectively. Here, we uncover redundant and specific roles of STAG1 and STAG2 in gene regulation and chromatin looping using HCT116 cells with an auxin-inducible degron (AID) tag fused to either STAG1 or STAG2. Following rapid depletion of either subunit, we perform high-resolution Hi-C, gene expression, and sequential ChIP studies to show that STAG1 and STAG2 do not co-occupy in- dividual binding sites and have distinct ways by which they affect looping and gene expression.
    [Show full text]
  • Upon Microbial Challenge, Human Neutrophils Undergo Rapid Changes in Nuclear Architecture and Chromatin Folding to Orchestrate an Immediate Inflammatory Gene Program
    Downloaded from genesdev.cshlp.org on October 5, 2021 - Published by Cold Spring Harbor Laboratory Press Upon microbial challenge, human neutrophils undergo rapid changes in nuclear architecture and chromatin folding to orchestrate an immediate inflammatory gene program Matthew Denholtz,1,5 Yina Zhu,1,5 Zhaoren He,1 Hanbin Lu,1 Takeshi Isoda,1,4 Simon Döhrmann,2 Victor Nizet,2,3 and Cornelis Murre1 1Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92039, USA; 2Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, California 92093, USA; 3Skaggs School of Pharmaceutical Sciences, University of California at San Diego, La Jolla, California 92093, USA Differentiating neutrophils undergo large-scale changes in nuclear morphology. How such alterations in structure are established and modulated upon exposure to microbial agents is largely unknown. Here, we found that prior to encounter with bacteria, an armamentarium of inflammatory genes was positioned in a transcriptionally passive environment suppressing premature transcriptional activation. Upon microbial exposure, however, human neu- trophils rapidly (<3 h) repositioned the ensemble of proinflammatory genes toward the transcriptionally permissive compartment. We show that the repositioning of genes was closely associated with the swift recruitment of cohesin across the inflammatory enhancer landscape, permitting an immediate transcriptional response upon bacterial exposure. We found that activated enhancers, marked by increased deposition of H3K27Ac, were highly enriched for cistromic elements associated with PU.1, CEBPB, TFE3, JUN, and FOSL2 occupancy. These data reveal how upon microbial challenge the cohesin machinery is recruited to an activated enhancer repertoire to instruct changes in chromatin folding, nuclear architecture, and to activate an inflammatory gene program.
    [Show full text]
  • Structure of the Human Cohesin Inhibitor Wapl
    Structure of the human cohesin inhibitor Wapl Zhuqing Ouyanga,1, Ge Zhenga,1, Jianhua Songb, Dominika M. Borekc, Zbyszek Otwinowskic, Chad A. Brautigamc, Diana R. Tomchickc, Susannah Rankinb, and Hongtao Yua,2 aHoward Hughes Medical Institute, Department of Pharmacology, and cDepartment of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390; and bProgram in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 Edited by Douglas Koshland, University of California, Berkeley, CA, and approved May 23, 2013 (received for review March 11, 2013) Cohesin, along with positive regulators, establishes sister-chromatid trigger cohesin release from chromosome arms (15, 18–21). A pool of cohesion by forming a ring to circle chromatin. The wings apart-like cohesin at centromeres is protected by the shugoshin (Sgo1)–protein protein (Wapl) is a key negative regulator of cohesin and forms phosphatase 2A (PP2A) complex (22, 23), which binds to cohesin, a complex with precocious dissociation of sisters protein 5 (Pds5) to dephosphorylates sororin, and protects cohesin from Wapl promote cohesin release from chromatin. Here we report the at centromeres (24). After all sister kinetochores attach properly crystal structure and functional characterization of human Wapl. to the mitotic spindle and are under tension, separase cleaves Wapl contains a flexible, variable N-terminal region (Wapl-N) and centromeric cohesin to initiate sister-chromatid separation. The a conserved C-terminal domain (Wapl-C) consisting of eight HEAT separated chromatids are evenly partitioned into the two daughter (Huntingtin, Elongation factor 3, A subunit, and target of rapamycin) cells through their attachment to microtubules originating from the repeats.
    [Show full text]
  • Ultrasound-Assisted Nonviral Gene Transfer of AQP1 to the Irradiated Minipig Parotid Gland Restores fluid Secretion
    Gene Therapy (2015) 22, 739–749 © 2015 Macmillan Publishers Limited All rights reserved 0969-7128/15 www.nature.com/gt ORIGINAL ARTICLE Ultrasound-assisted nonviral gene transfer of AQP1 to the irradiated minipig parotid gland restores fluid secretion Z Wang1, L Zourelias1,CWu1, PC Edwards2, M Trombetta3 and MJ Passineau1 Xerostomia is a common side effect of ionizing radiation used to treat head and neck cancer. A groundbreaking Phase I human clinical trial using Adenoviral gene transfer of Aquaporin-1 (AQP1) to a single salivary gland of individuals suffering from radiation- induced xerostomia has recently been reported. Unfortunately, the limitations of the Adenoviral vector system used in this pioneering trial preclude its advancement to a Phase II trial, and we have thus undertaken to evaluate the therapeutic potential of ultrasound-assisted nonviral gene transfer (UAGT) as an alternative means of delivering AQP1 gene therapy to the salivary gland by comparing head-to-head with the canonical Adenoviral vector in a swine model. Swine irradiated unilaterally with a 10-Gy electron beam targeted at the parotid gland suffered from significant, sustained hyposalivation that was bilateral, despite irradiation being confined to the targeted gland. Unilateral AQP1 gene therapy with UAGT resulted in bilateral restoration of stimulated salivary flow at 48 h and 1 week post treatment (1.62 ± 0.48 ml and 1.87 ± 0.45 ml) to preinjury levels (1.34 ± 0.14 ml) in a manner comparable to Adenoviral delivery (2.32 ± 0.6 ml and 1.33 ± 0.97 ml). UAGT can replace the Adenoviral vector as a means of delivering AQP1 gene therapy in the irradiated swine model, and it is a candidate for advancement to a Phase I human clinical trial.
    [Show full text]
  • A Requirement for STAG2 in Replication Fork Progression Creates a Targetable Synthetic Lethality in Cohesin-Mutant Cancers
    ARTICLE https://doi.org/10.1038/s41467-019-09659-z OPEN A requirement for STAG2 in replication fork progression creates a targetable synthetic lethality in cohesin-mutant cancers Gourish Mondal1, Meredith Stevers1, Benjamin Goode 1, Alan Ashworth2,3 & David A. Solomon 1,2 Cohesin is a multiprotein ring that is responsible for cohesion of sister chromatids and formation of DNA loops to regulate gene expression. Genomic analyses have identified that 1234567890():,; the cohesin subunit STAG2 is frequently inactivated by mutations in cancer. However, the reason STAG2 mutations are selected during tumorigenesis and strategies for therapeutically targeting mutant cancer cells are largely unknown. Here we show that STAG2 is essential for DNA replication fork progression, whereby STAG2 inactivation in non-transformed cells leads to replication fork stalling and collapse with disruption of interaction between the cohesin ring and the replication machinery as well as failure to establish SMC3 acetylation. As a con- sequence, STAG2 mutation confers synthetic lethality with DNA double-strand break repair genes and increased sensitivity to select cytotoxic chemotherapeutic agents and PARP or ATR inhibitors. These studies identify a critical role for STAG2 in replication fork procession and elucidate a potential therapeutic strategy for cohesin-mutant cancers. 1 Department of Pathology, University of California, San Francisco, CA 94143, USA. 2 UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA. 3 Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA 94158, USA. Correspondence and requests for materials should be addressed to D.A.S. (email: [email protected]) NATURE COMMUNICATIONS | (2019) 10:1686 | https://doi.org/10.1038/s41467-019-09659-z | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-09659-z ohesin is a multi-protein complex composed of four core transcriptional dysregulation22,23.
    [Show full text]
  • Loss of Cohesin Complex Components STAG2 Or STAG3 Confers Resistance to BRAF Inhibition in Melanoma
    Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Shen, C., S. H. Kim, S. Trousil, D. T. Frederick, A. Piris, P. Yuan, L. Cai, et al. 2016. “Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma.” Nature medicine 22 (9): 1056-1061. doi:10.1038/nm.4155. http:// dx.doi.org/10.1038/nm.4155. Published Version doi:10.1038/nm.4155 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:31731818 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nat Med Manuscript Author . Author manuscript; Manuscript Author available in PMC 2017 March 01. Published in final edited form as: Nat Med. 2016 September ; 22(9): 1056–1061. doi:10.1038/nm.4155. Loss of cohesin complex components STAG2 or STAG3 confers resistance to BRAF inhibition in melanoma Che-Hung Shen1, Sun Hye Kim1, Sebastian Trousil1, Dennie T. Frederick2, Adriano Piris3, Ping Yuan1, Li Cai1, Lei Gu4, Man Li1, Jung Hyun Lee1, Devarati Mitra1, David E. Fisher1,2, Ryan J. Sullivan2, Keith T. Flaherty2, and Bin Zheng1,* 1Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 2Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA 3Department of Dermatology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 4Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA.
    [Show full text]
  • Low Tolerance for Transcriptional Variation at Cohesin Genes Is Accompanied by Functional Links to Disease-Relevant Pathways
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.11.037358; this version posted April 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Title Low tolerance for transcriptional variation at cohesin genes is accompanied by functional links to disease-relevant pathways Authors William Schierdingǂ1, Julia Horsfieldǂ2,3, Justin O’Sullivan1,3,4 ǂTo whom correspondence should be addressed. 1 Liggins Institute, The University of Auckland, Auckland, New Zealand 2 Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand 3 The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Auckland, New Zealand 4 MRC Lifecourse Epidemiology Unit, University of Southampton Acknowledgements This work was supported by a Royal Society of New Zealand Marsden Grant to JH and JOS (16-UOO- 072), and WS was supported by the same grant. Contributions WS planned the study, performed analyses, and drafted the manuscript. JH and JOS revised the manuscript. Competing interests None declared. bioRxiv preprint doi: https://doi.org/10.1101/2020.04.11.037358; this version posted April 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Variants in DNA regulatory elements can alter the regulation of distant genes through spatial- regulatory connections.
    [Show full text]
  • Glioblastoma Cells Containing Mutations in the Cohesin Component STAG2 Are Sensitive to PARP Inhibition
    Published OnlineFirst December 19, 2013; DOI: 10.1158/1535-7163.MCT-13-0749 Molecular Cancer Cancer Biology and Signal Transduction Therapeutics Glioblastoma Cells Containing Mutations in the Cohesin Component STAG2 Are Sensitive to PARP Inhibition Melanie L. Bailey1, Nigel J. O'Neil1, Derek M. van Pel2, David A. Solomon3, Todd Waldman4, and Philip Hieter1 Abstract Recent data have identified STAG2, a core subunit of the multifunctional cohesin complex, as a highly recurrently mutated gene in several types of cancer. We sought to identify a therapeutic strategy to selectively target cancer cells harboring inactivating mutations of STAG2 using two independent pairs of isogenic glioblastoma cell lines containing either an endogenous mutant STAG2 allele or a wild-type STAG2 allele restored by homologous recombination. We find that mutations in STAG2 are associated with significantly increased sensitivity to inhibitors of the DNA repair enzyme PARP. STAG2-mutated, PARP-inhibited cells accumulated in G2 phase and had a higher percentage of micronuclei, fragmented nuclei, and chromatin bridges compared with wild-type STAG2 cells. We also observed more 53BP1 foci in STAG2-mutated glioblastoma cells, suggesting that these cells have defects in DNA repair. Furthermore, cells with mutations in STAG2 were more sensitive than cells with wild-type STAG2 when PARP inhibitors were used in combination with DNA-damaging agents. These data suggest that PARP is a potential target for tumors harboring inactivating mutations in STAG2, and strongly recommend that STAG2 status be determined and correlated with therapeutic response to PARP inhibitors, both prospectively and retrospectively, in clinical trials. Mol Cancer Ther; 13(3); 724–32.
    [Show full text]
  • Lecture9'21 Chromatin II
    Genetic Organization -Chromosomal Arrangement: From Form to Function. Chapters 9 & 10 in Genes XI The Eukaryotic chromosome – Organized Structures -banding – Centromeres – Telomeres – Nucleosomes – Euchromatin / Heterochromatin – Higher Orders of Chromosomal Structure 2 Heterochromatin differs from euchromatin in that heterochromatin is effectively inert; remains condensed during interphase; is transcriptionally repressed; replicates late in S phase and may be localized to the centromere or nuclear periphery Facultative heterochromatin is not restricted by pre-designated sequence; genes that are moved within or near heterochromatic regions can become inactivated as a result of their new location. Heterochromatin differs from euchromatin in that heterochromatin is effectively inert; remains condensed during interphase; is transcriptionally repressed; replicates late in S phase and may be localized to the centromere or nuclear periphery Facultative heterochromatin is not restricted by pre-designated sequence; genes that are moved within or near heterochromatic regions can become inactivated as a result of their new location. Chromatin inactivation (or heterochromatin formation) occurs by the addition of proteins to the nucleosomal fiber. May be due to: Chromatin condensation -making it inaccessible to transcriptional apparatus Proteins that accumulate and inhibit accessibility to the regulatory sequences Proteins that directly inhibit transcription Chromatin Is Fundamentally Divided into Euchromatin and Heterochromatin • Individual chromosomes can be seen only during mitosis. • During interphase, the general mass of chromatin is in the form of euchromatin, which is slightly less tightly packed than mitotic chromosomes. TF20210119 Regions of compact heterochromatin are clustered near the nucleolus and nuclear membrane Photo courtesy of Edmund Puvion, Centre National de la Recherche Scientifique Chromatin: Basic Structures • nucleosome – The basic structural subunit of chromatin, consisting of ~200 bp of DNA wrapped around an octamer of histone proteins.
    [Show full text]
  • THE DEVELOPMENT of CHEMICAL METHODS to DISCOVER KINASE SUBSTRATES and MAP CELL SIGNALING with GAMMA-MODIFIED ATP ANALOG-DEPENDENT KINASE-CATALYZED PHOSPHORYLATION By
    Wayne State University Wayne State University Dissertations 1-1-2017 The evelopmeD nt Of Chemical Methods To Discover Kinase Substrates And Map Cell Signaling With Gamma-Modified Atp Analog- Dependent Kinase-Catalyzed Phosphorylation Dissanayaka Mudiyanselage Maheeka Madhubashini Embogama Wayne State University, Follow this and additional works at: https://digitalcommons.wayne.edu/oa_dissertations Part of the Analytical Chemistry Commons, and the Biochemistry Commons Recommended Citation Embogama, Dissanayaka Mudiyanselage Maheeka Madhubashini, "The eD velopment Of Chemical Methods To Discover Kinase Substrates And Map Cell Signaling With Gamma-Modified Atp Analog-Dependent Kinase-Catalyzed Phosphorylation" (2017). Wayne State University Dissertations. 1698. https://digitalcommons.wayne.edu/oa_dissertations/1698 This Open Access Dissertation is brought to you for free and open access by DigitalCommons@WayneState. It has been accepted for inclusion in Wayne State University Dissertations by an authorized administrator of DigitalCommons@WayneState. THE DEVELOPMENT OF CHEMICAL METHODS TO DISCOVER KINASE SUBSTRATES AND MAP CELL SIGNALING WITH GAMMA-MODIFIED ATP ANALOG-DEPENDENT KINASE-CATALYZED PHOSPHORYLATION by DISSANAYAKA M. MAHEEKA M. EMBOGAMA DISSERTATION Submitted to the Graduate School of Wayne State University, Detroit, Michigan in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY 2017 MAJOR: CHEMISTRY (Biochemistry) Approved By: Advisor Date DEDICATION To my beloved mother, father, husband, daughter and sister. ii ACKNOWLEGEMENTS Many people have helped me during the past five years of earning my PhD. I would like to take this opportunity to convey my gratitude to them. First and foremost, I would like to thank my research supervisor Dr. Mary Kay Pflum for being the greatest mentor that I have met so far.
    [Show full text]